Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Dev Cell ; 59(1): 91-107.e6, 2024 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-38091997

RESUMEN

Genomic regulation of cardiomyocyte differentiation is central to heart development and function. This study uses genetic loss-of-function human-induced pluripotent stem cell-derived cardiomyocytes to evaluate the genomic regulatory basis of the non-DNA-binding homeodomain protein HOPX. We show that HOPX interacts with and controls cardiac genes and enhancer networks associated with diverse aspects of heart development. Using perturbation studies in vitro, we define how upstream cell growth and proliferation control HOPX transcription to regulate cardiac gene programs. We then use cell, organoid, and zebrafish regeneration models to demonstrate that HOPX-regulated gene programs control cardiomyocyte function in development and disease. Collectively, this study mechanistically links cell signaling pathways as upstream regulators of HOPX transcription to control gene programs underpinning cardiomyocyte identity and function.


Asunto(s)
Células Madre Pluripotentes Inducidas , Miocitos Cardíacos , Animales , Humanos , Miocitos Cardíacos/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Pez Cebra/metabolismo , Diferenciación Celular/genética , Proliferación Celular
2.
Zebrafish ; 19(1): 7-17, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35108124

RESUMEN

Heme oxygenase function is highly conserved between vertebrates where it plays important roles in normal embryonic development and controls oxidative stress. Expression of the zebrafish heme oxygenase 1 genes is known to be responsive to oxidative stress suggesting a conserved physiological function. In this study, we generate a knockout allele of zebrafish hmox1a and characterize the effects of hmox1a and hmox1b loss on embryonic development. We find that loss of hmox1a or hmox1b causes developmental defects in only a minority of embryos, in contrast to Hmox1 gene deletions in mice that cause loss of most embryos. Using a tail wound inflammation assay we find a conserved role for hmox1a, but not hmox1b, in normal macrophage migration to the wound site. Together our results indicate that zebrafish hmox1a has clearly a partitioned role from hmox1b that is more consistent with conserved functions of mammalian Heme oxygenase 1.


Asunto(s)
Hemo Oxigenasa (Desciclizante) , Pez Cebra , Animales , Hemo Oxigenasa (Desciclizante)/metabolismo , Hemo Oxigenasa (Desciclizante)/farmacología , Macrófagos/metabolismo , Ratones , Estrés Oxidativo , Pez Cebra/metabolismo
3.
Cytotherapy ; 23(12): 1074-1084, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34588150

RESUMEN

BACKGROUND AIMS: Mesenchymal stromal cells (MSCs) have been shown to improve cardiac function after injury and are the subject of ongoing clinical trials. In this study, the authors tested the cardiac regenerative potential of an induced pluripotent stem cell-derived MSC (iPSC-MSC) population (Cymerus MSCs) in a rat model of myocardial ischemia-reperfusion (I/R). Furthermore, the authors compared this efficacy with bone marrow-derived MSCs (BM-MSCs), which are the predominant cell type in clinical trials. METHODS: Four days after myocardial I/R injury, rats were randomly assigned to (i) a Cymerus MSC group (n = 15), (ii) a BM-MSC group (n = 15) or (iii) a vehicle control group (n = 14). For cell-treated animals, a total of 5 × 106 cells were injected at three sites within the infarcted left ventricular (LV) wall. RESULTS: One month after cell transplantation, Cymerus MSCs improved LV function (assessed by echocardiography) compared with vehicle and BM-MSCs. Interestingly, Cymerus MSCs enhanced angiogenesis without sustained engraftment or significant impact on infarct scar size. Suggesting safety, Cymerus MSCs had no effect on inducible tachycardia or the ventricular scar heterogeneity that provides a substrate for cardiac re-entrant circuits. CONCLUSIONS: The authors here demonstrate that intra-myocardial administration of iPSC-MSCs (Cymerus MSCs) provide better therapeutic effects compared with conventional BM-MSCs in a rodent model of myocardial I/R. Because of its manufacturing scalability, iPSC-MSC therapy offers an exciting opportunity for an "off-the-shelf" stem cell therapy for cardiac repair.


Asunto(s)
Células Madre Pluripotentes Inducidas , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Infarto del Miocardio , Animales , Infarto del Miocardio/terapia , Miocardio , Ratas
4.
Int J Cardiol ; 341: 24-30, 2021 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-34265313

RESUMEN

BACKGROUND: Novel therapies that can limit or reverse damage caused by myocardial infarction (MI) could ease the increasing burden of heart failure. In this regard Platelet Derived Growth Factor (PDGF) has been previously shown to contribute to cardiac repair after MI. Here, we use a rodent model of MI and recombinant adeno-associated virus 9 (rAAV9)-mediated gene transfer to overexpress Pdgf-a in the injured heart and assess its therapeutic potential. METHODS AND RESULTS: Sprague Dawley rats underwent temporary occlusion of the left anterior descending coronary artery, followed immediately by systemic delivery of 1 × 10^11 vector genomes of either rAAV9 Pdgf-a or rAAV9 Empty vector (control). At day 28 post-MI echocardiography showed significantly improved left ventricular (LV) function (fractional shortening) after rAAV9 Pdgf-a (0.394 ± 0.019%) treatment vs control (0.304 ± 0.018%). Immunohistochemical analysis demonstrated significantly increased capillary and arteriolar density in the infarct border zone of rAAV9 Pdgf-a treated hearts together with a significant reduction in infarct scar size (rAAV9 Pdgf-a 6.09 ± 0.94% vs Empty 12.45 ± 0.92%). Western blot and qPCR analyses confirmed overexpression of PDGF-A and showed upregulation of smooth muscle alpha actin (Acta2), collagen type III alpha 1 (Col3a1) and lysyl oxidase (Lox) genes in rAAV9 Pdgf-a treated infarcts. CONCLUSION: Overexpression of Pdgf-a in the post-MI heart can modulate scar composition and improve LV function. Our study highlights the potential of rAAV gene transfer of Pdgf-a as a cardio-reparative therapy.


Asunto(s)
Cicatriz , Infarto del Miocardio , Animales , Modelos Animales de Enfermedad , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/genética , Infarto del Miocardio/terapia , Miocardio/patología , Factor de Crecimiento Derivado de Plaquetas/genética , Ratas , Ratas Sprague-Dawley , Función Ventricular Izquierda , Remodelación Ventricular
5.
Science ; 372(6538): 201-205, 2021 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-33833125

RESUMEN

Cardiac regeneration requires dedifferentiation and proliferation of mature cardiomyocytes, but the mechanisms underlying this plasticity remain unclear. Here, we identify a potent cardiomyogenic role for Krüppel-like factor 1 (Klf1/Eklf), which is induced in adult zebrafish myocardium upon injury. Myocardial inhibition of Klf1 function does not affect heart development, but it severely impairs regeneration. Transient Klf1 activation is sufficient to expand mature myocardium in uninjured hearts. Klf1 directs epigenetic reprogramming of the cardiac transcription factor network, permitting coordinated cardiomyocyte dedifferentiation and proliferation. Myocardial expansion is supported by Klf1-induced rewiring of mitochondrial metabolism from oxidative respiration to anabolic pathways. Our findings establish Klf1 as a core transcriptional regulator of cardiomyocyte renewal in adult zebrafish hearts.


Asunto(s)
Reprogramación Celular , Corazón/fisiología , Factores de Transcripción de Tipo Kruppel/metabolismo , Miocitos Cardíacos/fisiología , Regeneración , Proteínas de Pez Cebra/metabolismo , Animales , Cardiomegalia Inducida por el Ejercicio , Desdiferenciación Celular , Diferenciación Celular , Proliferación Celular , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Glucólisis , Corazón/embriología , Ventrículos Cardíacos/citología , Factores de Transcripción de Tipo Kruppel/genética , Desarrollo de Músculos , Miocardio/metabolismo , Miocitos Cardíacos/citología , Vía de Pentosa Fosfato , Pez Cebra , Proteínas de Pez Cebra/genética
6.
Heart Lung Circ ; 27(8): 967-975, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29573957

RESUMEN

BACKGROUND: Vitamin D has been implicated in the prevention of heart failure. However the underlying mechanism remains unclear. We hypothesised that these effects may be partially mediated by cardiac stem/progenitor cells (CPCs). Therefore, we examined the effects of 1,25-dihydroxyvitamin D3 (1,25D) on cell cycle activity and differentiation of a previously described CPC population called cardiac colony-forming unit fibroblasts (cCFU-Fs). METHODS: cCFU-Fs were isolated from adult male C57Bl/6 mouse hearts using fluorescence-activated cell sorting. The effect of 1,25D on cell proliferation and differentiation were was assessed by colony-forming and fibroblast differentiation assays. Cell cycle was analysed by flow cytometry. Mice with induced myocardial infarction (MI) were treated with 1,25D or vehicle controls and cardiac function assessed by echocardiography. RESULTS: 1,25D dose-dependently increased expression of vitamin D receptor (Vdr) and reduced large colony formation. Addition of 1,25D to cCFU-Fs slowed cell proliferation, promoted cell cycle arrest and decreased expression of pro-fibrotic factors during TGF-ß-induced fibroblast differentiation of cCFU-Fs. After MI, 1,25D-treated mice had less left ventricular wall thinning and significant improvement in left ventricular systolic function compared to vehicle-treated controls. Although no significant changes in myocardial fibrotic area and cardiomyocyte size were noted, treatment with 1,25D significantly inhibited cardiac interstitial cell proliferation after MI. CONCLUSIONS: Vitamin D signalling promotes cardioprotection after myocardial infarction. This may be through modulation of cCFU-F cell cycle. The role of 1,25D and VDR in regulating cardiac stem/progenitor cell function therefore warrants further investigation.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Insuficiencia Cardíaca/prevención & control , Infarto del Miocardio/terapia , Miocitos Cardíacos/citología , Células Madre/citología , Vitamina D/uso terapéutico , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Ecocardiografía , Citometría de Flujo , Insuficiencia Cardíaca/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/diagnóstico , Vitaminas/uso terapéutico
7.
Hypertens Res ; 39(11): 764-768, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27334059

RESUMEN

Chronic hypertension causes vascular remodeling that is associated with an increase in periostin- (postn) positive cells, including fibroblasts and smooth muscle cells. Krüppel-like factor (KLF) 5, a transcription factor, is also observed in vascular remodeling; however, it is unknown what role KLF5 plays in postn-positive cells during vascular remodeling induced by deoxycorticosterone-acetate (DOCA) salt. We used postn-positive cell-specific Klf5-deficient mice (Klf5PostnKO: Klf5flox/flox; PostnCre/-) and wild-type mice (WT: Klf5flox/flox; Postn-/-). We implanted a DOCA pellet and provided drinking water containing 0.9% NaCl for 8 weeks. The DOCA-salt treatment induced hypertension in both genotypes, as observed by increases in systolic blood pressure. In WT animals, DOCA-salt treatment increased the aortic medial area compared with the non-treated controls. Similarly, Tgfb1 was overexpressed in the aortas of the DOCA-salt treated WT mice compared with the controls. Immunofluorescence staining revealed that fibroblast-specific protein 1 (FSP1)+-α smooth muscle actin (αSMA)+ myofibroblasts exist in the medial area of the WT aortas after DOCA-salt intervention. Importantly, these changes were not observed in the Klf5PostnKO animals. In conclusion, the results of this study suggest that the presence of KLF5 in postn-positive cells contributes to the pathogenesis of aortic thickening induced by DOCA-salt hypertension.


Asunto(s)
Aorta/metabolismo , Moléculas de Adhesión Celular/metabolismo , Hipertensión/genética , Factores de Transcripción de Tipo Kruppel/genética , Remodelación Vascular/genética , Animales , Presión Sanguínea/fisiología , Acetato de Desoxicorticosterona , Hipertensión/inducido químicamente , Hipertensión/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Ratones Noqueados , Miofibroblastos/metabolismo , Proteína de Unión al Calcio S100A4/genética , Proteína de Unión al Calcio S100A4/metabolismo
8.
J Cardiol ; 68(3): 253-60, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26475067

RESUMEN

BACKGROUND: The renin angiotensin system (RAS) plays an important role in the pathogenesis of cardiovascular diseases and inflammation. Myocarditis is an inflammatory disease of the heart, and the role of the RAS in its pathophysiology is unknown. Because the direct renin inhibitor, aliskiren, is thought to block RAS completely, we investigated the cardioprotective effect of aliskiren in mice with experimental autoimmune myocarditis (EAM). METHODS: A cardiac α-myosin heavy chain peptide was injected in mice on days 0 and 7. Aliskiren 25mg/kg per day (n=10) or vehicle (n=10) was administered to EAM mice starting on day 0 and the animals were killed on day 21. RESULTS: Aliskiren significantly prevented the progression of left ventricular wall thickening in EAM hearts compared to the vehicle-treated group. Histologically, the inflammatory cell infiltration and fibrosis area ratios in the aliskiren-treated group were lower than that in the vehicle-treated group. Immunohistochemistry revealed that aliskiren suppressed CD4 positive cell infiltration in EAM hearts compared to vehicle. Moreover, aliskiren decreased mRNA levels of interleukin (IL)-2, interferon-γ, tumor necrosis factor-α, and collagen 1. In vitro study showed that aliskiren inhibited T cell proliferation and IL-2 production induced by myosin stimulation. CONCLUSION: Our results suggest that aliskiren ameliorates EAM by suppressing T-cell activation and inflammatory cytokines, and has potential as a treatment for myocarditis.


Asunto(s)
Amidas/farmacología , Antihipertensivos/farmacología , Fumaratos/farmacología , Enfermedad Autoinmune Experimental del Sistema Nervioso/tratamiento farmacológico , Renina/antagonistas & inhibidores , Animales , Citocinas/genética , Citocinas/metabolismo , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo
9.
J Appl Genet ; 57(1): 91-7, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26025227

RESUMEN

The role of angiotensin II type 1 (AT1) receptors in muscle development and hypertrophy remains unclear. This study was designed to reveal the effects that a loss of AT1 receptors has on skeletal muscle development and hypertrophy in mice. Eight-week-old male AT1a receptor knockout (AT1a(-/-)) mice were used for this experiment. The plantaris muscle to body weight ratio, muscle fiber cross-sectional area, and number of muscle fibers of AT1a(-/-) mice was significantly greater than wild type (WT) mice in the non-intervention condition. Next, the functional overload (OL) model was used to induce plantaris muscle hypertrophy by surgically removing the two triceps muscles consisting of the calf, soleus, and gastrocnemius muscles in mice. After 14 days of OL intervention, the plantaris muscle weight, the amount of fiber, and the fiber area increased. However, the magnitude of the increment of plantaris weight was not different between the two strains. Agtr1a mRNA expression did not change after OL in WT muscle. Actually, the Agt mRNA expression level of WT-OL was lower than WT-Control (C) muscle. An atrophy-related gene, atrogin-1 mRNA expression levels of AT1a(-/-)-C, WT-OL, and AT1a(-/-)-OL muscle were lower than that of WT-C muscle. Our findings suggest that AT1 receptor contributes to plantaris muscle development via atrogin-1 in mice.


Asunto(s)
Hipertrofia/genética , Músculo Esquelético/crecimiento & desarrollo , Receptor de Angiotensina Tipo 1/metabolismo , Angiotensina I/sangre , Angiotensina II/sangre , Animales , Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Musculares/metabolismo , ARN Mensajero/metabolismo , Receptor de Angiotensina Tipo 1/genética , Proteínas Ligasas SKP Cullina F-box/metabolismo
10.
PLoS One ; 10(3): e0119360, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25768281

RESUMEN

Myocarditis is a critical inflammatory disorder which causes life-threatening conditions. No specific or effective treatment has been established. DPP-4 inhibitors have salutary effects not only on type 2 diabetes but also on certain cardiovascular diseases. However, the role of a DPP-4 inhibitor on myocarditis has not been investigated. To clarify the effects of a DPP-4 inhibitor on myocarditis, we used an experimental autoimmune myocarditis (EAM) model in Balb/c mice. EAM mice were assigned to the following groups: EAM mice group treated with a DPP-4 inhibitor (linagliptin) (n = 19) and those untreated (n = 22). Pathological analysis revealed that the myocardial fibrosis area ratio in the treated group was significantly lower than in the untreated group. RT-PCR analysis demonstrated that the levels of mRNA expression of IL-2, TNF-α, IL-1ß and IL-6 were significantly lower in the treated group than in the untreated group. Lymphocyte proliferation assay showed that treatment with the DPP-4 inhibitor had no effect on antigen-induced spleen cell proliferation. Administration of the DPP-4 inhibitor remarkably suppressed cardiac fibrosis and reduced inflammatory cytokine gene expression in EAM mice. Thus, the agents present in DPP-4 inhibitors may be useful to treat and/or prevent clinical myocarditis.


Asunto(s)
Enfermedades Autoinmunes/patología , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Linagliptina/farmacología , Miocarditis/patología , Animales , Enfermedades Autoinmunes/tratamiento farmacológico , Enfermedades Autoinmunes/genética , Proliferación Celular/efectos de los fármacos , Inhibidores de la Dipeptidil-Peptidasa IV/uso terapéutico , Modelos Animales de Enfermedad , Fibrosis , Corazón/efectos de los fármacos , Corazón/fisiopatología , Pruebas de Función Cardíaca , Inflamación/tratamiento farmacológico , Inflamación/genética , Inflamación/patología , Linagliptina/uso terapéutico , Pulmón/efectos de los fármacos , Pulmón/patología , Linfocitos/efectos de los fármacos , Linfocitos/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Miocarditis/tratamiento farmacológico , Miocarditis/genética , Tamaño de los Órganos/efectos de los fármacos
11.
Heart Vessels ; 30(4): 527-33, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24879505

RESUMEN

Myocarditis is a clinically serious disease; however, no effective treatment has been elucidated. The P2X7 receptor is related to the pathophysiology of inflammation in many cardiovascular diseases. The P2X7 receptor antagonist is promising as an immunosuppressive treatment; however, its role in myocarditis is still to be established. To clarify the role of the P2X7 receptor, we used a murine experimental autoimmune myocarditis (EAM) model. Mice were immunized on day 0 and 7 with synthetic cardiac myosin peptide to establish EAM. The mice with induced EAM were treated with A740003, the P2X7 receptor antagonist (n = 10), or not treated (n = 11); hearts were harvested on day 21. The P2X7 receptor antagonist improved myocardial contraction of the EAM hearts via suppressed infiltration of CD4+ T cells and macrophages. Similarly, mRNA expression of interleukin 1 beta, the P2X7 receptor and NADPH oxidase 2/4 was lower in the heart of the P2X7 receptor antagonist-treated group compared to the non-treat group. The P2X7 receptor antagonist suppressed EAM development; thus, this inhibition is promising for treating clinical myocarditis.


Asunto(s)
Acetamidas/uso terapéutico , Enfermedades Autoinmunes/tratamiento farmacológico , Miocarditis/tratamiento farmacológico , NADPH Oxidasas/metabolismo , Antagonistas del Receptor Purinérgico P2X/uso terapéutico , Quinolinas/uso terapéutico , Animales , Modelos Animales de Enfermedad , Inflamación/metabolismo , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Miocardio/patología , Receptores Purinérgicos P2X7/metabolismo
12.
Heart Vessels ; 29(4): 542-9, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24002697

RESUMEN

We previously demonstrated that Porphyromonas gingivalis infection induces neointimal hyperplasia with an increase in monocyte chemoattractant protein (MCP)-1 after arterial injury in wild-type mice. Toll-like receptor (TLR) 2 is a key receptor for the virulence factors of P. gingivalis. The aim of this study was to assess whether TLR2 plays a role in periodontopathic bacteria-induced neointimal formation after an arterial injury. Wild-type and TLR2-deficient mice were used in this study. The femoral arteries were injured, and P. gingivalis or vehicle was injected subcutaneously once per week. Fourteen days after arterial injury, the murine femoral arteries were obtained for histopathologic and immunohistochemical analyses. The immunoglobulin-G levels of the P. gingivalis-infected groups were significantly increased in comparison with the level in the corresponding noninfected groups in both wild-type and TLR2-deficient mice. TLR2 deficiency negated the P. gingivalis-induced neointimal formation in comparison with the wild-type mice, and reduced the number of positive monocyte chemoattractant protein-1 cells in the neointimal area. These findings demonstrate that P. gingivalis infection can promote neointimal formation after an arterial injury through TLR2 signaling.


Asunto(s)
Arteria Femoral/microbiología , Neointima , Porphyromonas gingivalis/patogenicidad , Transducción de Señal , Receptor Toll-Like 2/metabolismo , Lesiones del Sistema Vascular/microbiología , Animales , Anticuerpos Antibacterianos/sangre , Quimiocina CCL2/metabolismo , Modelos Animales de Enfermedad , Arteria Femoral/inmunología , Arteria Femoral/lesiones , Arteria Femoral/metabolismo , Arteria Femoral/patología , Hiperplasia , Inmunoglobulina G/sangre , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Porphyromonas gingivalis/inmunología , Receptor Toll-Like 2/deficiencia , Receptor Toll-Like 2/genética , Lesiones del Sistema Vascular/sangre , Lesiones del Sistema Vascular/genética , Lesiones del Sistema Vascular/inmunología , Lesiones del Sistema Vascular/patología
13.
Expert Opin Investig Drugs ; 23(2): 149-62, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24205804

RESUMEN

OBJECTIVE: Mechanisms of cardiac dysfunction in myocarditis have not been fully elucidated. Though it remains controversial whether angiogenesis is beneficial or harmful in inflammatory disease, significant vascular destruction might possibly impair cardiac function in myocarditis. The prolyl hydroxylase domain-containing protein (PHD) inhibitor is a potential drug for promoting angiogenesis as it stabilizes hypoxia inducible transcription factor (HIF). The authors examine whether the PHD inhibitor TM6008 could affect cardiac function by promoting angiogenesis in experimental autoimmune myocarditis (EAM). METHODS: EAM was induced on BALB/c mice by immunizing them with a synthesized α myosin heavy-chain peptide. Every day, 200 mg/kg of TM6008 or vehicle was administered orally. RESULTS: TM6008 improved left ventricular ejection fraction significantly on the 21st day of EAM. Though TM6008 did not affect the severity of myocardial cell infiltration, it tended to reduce cardiac fibrosis. Immunohistochemistry showed that CD31-positive blood vessels were preserved in the TM6008 group compared to the control group. Immunoblotting revealed that TM6008 increased the expression of HIF-1α, HIF-2α and vascular endothelial growth factor in myocarditis. CONCLUSION: Inhibition of PHD could ameliorate cardiac dysfunction in EAM, partially through promoting neovascularization. Relief of tissue hypoxia via neovascularization could improve cardiac function in myocarditis.


Asunto(s)
Enfermedades Autoinmunes/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Miocarditis/metabolismo , Inhibidores de Prolil-Hidroxilasa/farmacología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Enfermedades Autoinmunes/tratamiento farmacológico , Enfermedades Autoinmunes/fisiopatología , Vasos Coronarios/fisiología , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos BALB C , Miocarditis/tratamiento farmacológico , Miocarditis/fisiopatología , Cadenas Pesadas de Miosina/inmunología , Neovascularización Fisiológica , Inhibidores de Prolil-Hidroxilasa/uso terapéutico , Disfunción Ventricular Izquierda/tratamiento farmacológico , Disfunción Ventricular Izquierda/metabolismo , Disfunción Ventricular Izquierda/fisiopatología
14.
Int Heart J ; 54(6): 412-6, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24309453

RESUMEN

Recent reports assert that dental health is linked to an increased risk of cardiovascular disease. It is well known that Aggregatibacter actinomycetemcomitans (A.a.) is highly associated with heart disease. Indeed, we previously reported that A.a. affects the development of heart disease in a mouse model. However, no reports have clarified the relationship between A.a. and experimental autoimmune myocarditis (EAM). The aim of this study was to investigate the effects of A.a. on EAM in mice. EAM was induced via the injection of cardiac myosin into the mice. A.a. or PBS was then injected into the mice using a chamber implanted into the back of each mouse. The weight of the organs and echocardiograms were obtained and a pathological analysis and quantitative RT-PCR were performed. Echocardiography showed that no statistical difference was observed between the two groups. A histopathological analysis demonstrated that the number of areas affected by myocarditis in the A.a.-injected EAM group was significantly increased compared to that observed in the PBS-injected EAM group (P < 0.05). The hearts of the mice in the A.a.-injected EAM group exhibited significantly increased expressions of MMP-9 mRNA compared to the hearts of the mice in the PBS-injected EAM group (P < 0.05). These results show that A.a. aggravated EAM via an enhanced MMP expression.


Asunto(s)
Aggregatibacter actinomycetemcomitans , Enfermedades Autoinmunes/microbiología , Miocarditis/microbiología , Infecciones por Pasteurellaceae/complicaciones , Animales , Enfermedades Autoinmunes/metabolismo , Modelos Animales de Enfermedad , Masculino , Metaloproteinasa 9 de la Matriz/biosíntesis , Ratones , Ratones Endogámicos BALB C , Miocarditis/metabolismo
15.
Am J Physiol Heart Circ Physiol ; 305(12): H1761-71, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24097428

RESUMEN

NF-κB, which is activated by the inhibitor of NF-κB kinase (IKK), is involved in the progression of inflammatory disease. However, the effect of IKK inhibition on the progression of myocarditis is unknown. We examined the effect of IKK inhibition on the progression of myocarditis. Lewis rats were immunized with porcine cardiac myosin to induce experimental autoimmune myocarditis (EAM). We administered the IKK inhibitor (IMD-0354; 15 mg·kg(-1)·day(-1)) or vehicle to EAM rats daily. Hearts were harvested 21 days after immunization. Although the untreated EAM group showed increased heart weight-to-body weight ratio, and severe myocardial damage, these changes were attenuated in the IKK inhibitor-treated group. Moreover, IKK inhibitor administration significantly reduced NF-κB activation and mRNA expression of IFN-γ, IL-2, and monocyte chemoattractant protein-1 in myocardium compared with vehicle administration. In vitro study showed that the IKK inhibitor treatment inhibited T-cell proliferation and Th1 cytokines production induced by myosin stimulation. The IKK inhibitor ameliorated EAM by suppressing inflammatory reactions via suppression of T-cell activation.


Asunto(s)
Enfermedades Autoinmunes/tratamiento farmacológico , Benzamidas/uso terapéutico , Miocarditis/tratamiento farmacológico , FN-kappa B/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Linfocitos T/efectos de los fármacos , Animales , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Benzamidas/farmacología , Modelos Animales de Enfermedad , Masculino , Miocarditis/inmunología , Miocarditis/metabolismo , FN-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Ratas , Ratas Endogámicas Lew , Índice de Severidad de la Enfermedad , Linfocitos T/inmunología , Linfocitos T/metabolismo , Quinasa de Factor Nuclear kappa B
16.
Expert Opin Investig Drugs ; 22(9): 1095-106, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23915387

RESUMEN

BACKGROUND: Neointimal hyperplasia after the percutaneous coronary intervention is still a clinically serious problem, associated with the risk of thrombosis due to delayed reendothelization. Peroxisome proliferator-activated receptor-ß/δ (PPAR-ß/δ) belongs to a family of ligand-activated transcription factors. OBJECTIVES: In this study, we investigated the effects of GW-0742, a synthetic high-affinity PPAR-ß/δ agonist, on neointimal hyperplasia after arterial injury. Using C57BL/6J mice, we made a wire-injury model and intraperitoneally injected GW-0742 or vehicle once a day. The arteries were harvested for pathological and molecular analysis on day 14 after injury. In vitro, vascular smooth muscle cells (VSMCs), macrophages and human umbilical vein endothelial cells (HUVECs) were cultured, and GW-0742 effects on the cells proliferation were measured. RESULTS: The vehicle-treated injured arteries showed significantly thickened intima, while GW-0742 suppressed it. GW-0742 significantly suppressed IL-6 protein production, the expression of proliferating cell nuclear antigen in the neointima and enhanced CD31 expression. In vitro, GW-0742 attenuated VSMC proliferation triggered by cytokines or macrophages. The drug also induced endothelial regeneration after denudation injury. CONCLUSION: The data suggest that the PPAR-ß/δ agonist is effective for atten- uation of neointimal hyperplasia by suppressing VSMC proliferation and accelerating reendothelization.


Asunto(s)
Neointima/prevención & control , PPAR delta/agonistas , PPAR-beta/agonistas , Tiazoles/farmacología , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Hiperplasia/prevención & control , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , Neointima/patología , Regeneración/efectos de los fármacos
17.
Expert Opin Ther Targets ; 17(9): 1003-9, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23865521

RESUMEN

BACKGROUNDS: Salusins are multifunctional endogenous bioactive peptides simultaneously biosynthesized from their precursor prosalusin. Salusin-ß stimulates proliferation of vascular smooth muscle cells and fibroblasts and regulates myocardial growth and hypertrophy. Salusin-ß has potent hypotensive, bradycardic and proatherosclerotic effects. OBJECTIVES: To investigate whether salusin-ß plays a role in myocardial remodeling after myocardial ischemia reperfusion (I/R) injury, rat I/R models were created by the left anterior descending coronary artery occlusion for 30 min, followed by 24 h or 7 days of reperfusion (control, n = 6 each). RESULTS AND CONCLUSION: Immunohistochemical double staining showed the enhanced expression of salusin-ß in the macrophages around myocardial ischemic area. Anti-salusin-ß treated groups were administered the neutralizing salusin-ß antibody (10 µl/day, i.p.) once daily from day -1 to day 1 or from day -1 to day 7 (anti-salusin-ß, n = 6 each). The anti-salusin-ß therapy enhanced myocardial angiogenesis in the peri-ischemic area of reperfusion. The small vessels (< 40 µm in diameter) of I/R hearts treated with anti-salusin-ß were more densely populated than those of control animals (108.5 ± 19.7 vs 47.5 ± 2.4, p < 0.05). Real-time PCR revealed that the anti-salusin-ß therapy-induced angiogenesis was not associated with enhanced vascular endothelial growth factor A expression. The authors, for the first time, have clarified that endogenous salusin-ß suppresses angiogenesis which is critical in the development of cardiac remodeling following I/R injury.


Asunto(s)
Anticuerpos/uso terapéutico , Péptidos y Proteínas de Señalización Intercelular/inmunología , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Animales , Anticuerpos/farmacología , Línea Celular , Células Cultivadas , Quimiocina CCL2/genética , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Interleucina-8/genética , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/genética , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/fisiopatología , Miocardio/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Factor A de Crecimiento Endotelial Vascular/genética
18.
Int Heart J ; 54(3): 176-80, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23774243

RESUMEN

Chlorogenic acid (CGA), which is a key component of coffee, has many biological effects such as anti-inflammation activity. However, the effects of CGA on ventricular remodeling after myocardial ischemia have not been well investigated. To test the hypothesis that CGA can attenuate chronic ventricular remodeling after myocardial ischemia, we orally administered CGA to murine myocardial ischemia models. Seven to nine week-old C57BL/6 mice were used. A myocardial infarction (MI) model was produced by permanent ligation of the left anterior descending coronary artery (LAD) using an 8-0 suture passed under the arteries. These mice were randomly assigned into 4 groups in each experimental model. Some MI mice were supplemented orally with CGA (30 mg/kg/day, MI+CGA group, n = 13) as a CGAtreated MI group, and other MI mice received vehicle (MI+vehicle group, n = 11) as a vehicle-treated MI group. Shamoperated mice without MI also received vehicle (Sham+vehicle group, n = 3) as a sham group, and sham-operated mice without MI received CGA (30 mg/kg/day, Sham+CGA group, n = 8) as a Sham+CGA group. Just before sacrifice on day 14, we measured blood pressure and heart rate and performed echocardiography. We obtained 3 transverse sections per heart for histopathologic examination. There were no differences in body weight, heart rate, or blood pressure among the groups on day 14. The vehicle-treated MI group showed significantly impaired left ventricular contraction compared to the sham-operated group. However, the CGA-treated MI group showed significantly improved ventricular contraction compared to the vehicle-treated MI group. Severe myocardial fibrosis with enhanced macrophage infiltration was observed in the vehicle-treated ischemia group on day 14. CGA attenuated these fibrotic changes with suppressed macrophage infiltration without systemic adverse effects. CGA may effectively suppress chronic ventricular remodeling after myocardial ischemia because it is critically involved in the suppression of macrophage infiltration.


Asunto(s)
Ácido Clorogénico/administración & dosificación , Infarto del Miocardio/tratamiento farmacológico , Remodelación Ventricular/efectos de los fármacos , Animales , Ventrículos Cardíacos/fisiopatología , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL
19.
Circ J ; 77(6): 1565-73, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23412709

RESUMEN

BACKGROUND: Periodontopathic bacteria are detected at a high rate in specimens obtained from the aortic walls of patients with abdominal aortic aneurysm (AAA) and are involved in AAA development. The purpose of this study was to clarify the role of Toll-like receptors (TLRs), which are key receptors of virulence factors of many periodontal bacteria, on periodontopathic bacteria-accelerated AAA progression. METHODS AND RESULTS: AAA was produced by peri-aortic application of 0.25 mol/L CaCl2, with NaCl used as a control. The mice were inoculated with live Porphyromonas (P.) gingivalis or vehicle once weekly. At 4 weeks after the application of CaCl2, the aortic diameter of the P. gingivalis-infected wild-type mice showed a significant increase in comparison with vehicle control mice (P<0.05). The P. gingivalis-infected TLR-2 deficient mice showed no statistical increase in aortic diameter over the same period. The aortic diameter of the P. gingivalis-infected TLR-4 deficient mice statistically increased. Immunohistochemically, the levels of matrix metalloproteinase-2 and -9 in the aneurysmal samples from wild-type mice were higher than in TLR-2 deficient mice. CONCLUSIONS: P. gingivalis accelerated the progression of experimental AAA through TLR-2 signaling.


Asunto(s)
Aneurisma de la Aorta Abdominal , Infecciones por Bacteroidaceae , Periodoncio/microbiología , Porphyromonas gingivalis/inmunología , Transducción de Señal/inmunología , Receptor Toll-Like 2/inmunología , Animales , Aorta Abdominal/inmunología , Aorta Abdominal/microbiología , Aorta Abdominal/patología , Aneurisma de la Aorta Abdominal/genética , Aneurisma de la Aorta Abdominal/inmunología , Aneurisma de la Aorta Abdominal/microbiología , Aneurisma de la Aorta Abdominal/patología , Infecciones por Bacteroidaceae/genética , Infecciones por Bacteroidaceae/inmunología , Infecciones por Bacteroidaceae/patología , Humanos , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/inmunología , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/inmunología , Ratones , Ratones Noqueados , Periodoncio/inmunología , Periodoncio/patología , Transducción de Señal/genética , Receptor Toll-Like 2/genética , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología
20.
Am J Physiol Heart Circ Physiol ; 304(5): H740-8, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23262138

RESUMEN

Periodontitis is one of the most common infections in humans. Recently, published reports assert that periodontitis is associated with cardiovascular disease. Although it is said that viral, bacterial infections and autoimmune diseases may be the cause of myocarditis, the pathogenesis of it remains unclear. The aim of this study was to investigate the influence of a periodontal pathogen on experimental autoimmune myocarditis (EAM). Porphyromonas gingivalis (P.g.), PBS as a control, were injected into the mice. Histopathological and immunohistochemical analyses were performed. We examined heart mRNA levels using quantitative RT-PCR. The anti-P.g. IgG antibody level in plasma samples of the P.g.-injected group significantly increased compared with the PBS-injected group. Histopathological analysis detected that the myocarditis-affected areas and the fibrotic area in the P.g.-injected EAM group significantly increased compared with the PBS-injected EAM group (P < 0.05). Immunohistochemical analysis detected that more CD11b-positive cells were shown in the heart of the P.g.-injected EAM group compared with the PBS EAM-injected group (P < 0.05). Hearts from the P.g.-injected EAM group showed significantly increased expression of monocyte chemoattractant protein-1, IFN-γ, and matrix metalloproteinase-9 (MMP-9) mRNA compared with the hearts from the PBS-injected EAM group (P < 0.05). On day 7, serum levels of IL-6 were significantly enhanced in the P.g.-injected EAM group compared with the PBS-injected EAM group (P < 0.05). These results showed that P.g. injection could deteriorate EAM in mice through CD11b-positive cells, cytokines, and MMP-9 expression.


Asunto(s)
Anticuerpos Antibacterianos/sangre , Autoanticuerpos/sangre , Enfermedades Autoinmunes/inmunología , Miocarditis/inmunología , Periodontitis/inmunología , Porphyromonas gingivalis/inmunología , Animales , Enfermedades Autoinmunes/epidemiología , Enfermedades Autoinmunes/microbiología , Peso Corporal , Antígeno CD11b/inmunología , Citocinas/sangre , Citocinas/inmunología , Modelos Animales de Enfermedad , Pulmón/inmunología , Pulmón/patología , Masculino , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/inmunología , Ratones , Ratones Endogámicos BALB C , Miocarditis/epidemiología , Miocarditis/microbiología , Miosinas/inmunología , Tamaño de los Órganos/inmunología , Periodontitis/epidemiología , Periodontitis/microbiología , Prevalencia , Factores de Riesgo , Estudios Seroepidemiológicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA